Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Trends Immunol ; 41(8): 648-651, 2020 08.
Article in English | MEDLINE | ID: mdl-32622855

ABSTRACT

Multiple research groups have demonstrated that caspase-8 (CASP8)-mediated gasdermin D (GSDMD) cleavage drives pyroptotic cell death. Here, we discuss a novel role for the enzymatically inactive homolog of CASP8, the long isoform of cellular FLICE-like inhibitory protein (cFLIPL), in the regulation of this process. Specifically, cFLIP-deficiency provides a model in which to study the mechanisms regulating CASP8-mediated activation of cell death and inflammatory signaling.


Subject(s)
Apoptosis , Inflammation , Signal Transduction , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Caspase 8/metabolism , Humans , Inflammation/pathology , Models, Immunological , Pyroptosis , Signal Transduction/immunology
2.
Am J Physiol Gastrointest Liver Physiol ; 314(3): G319-G333, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29191940

ABSTRACT

Cholestatic liver injury results from impaired bile flow or metabolism and promotes hepatic inflammation and fibrogenesis. Toxic bile acids that accumulate in cholestasis induce apoptosis and contribute to early cholestatic liver injury, which is amplified by accompanying inflammation. The aim of the current study was to evaluate the role of the antiapoptotic caspase 8-homolog cellular FLICE-inhibitory (cFLIP) protein during acute cholestatic liver injury. Transgenic mice exhibiting hepatocyte-specific deletion of cFLIP (cFLIP-/-) were used for in vivo and in vitro analysis of cholestatic liver injury using bile duct ligation (BDL) and the addition of bile acids ex vivo. Loss of cFLIP in hepatocytes promoted acute cholestatic liver injury early after BDL, which was characterized by a rapid release of proinflammatory and chemotactic cytokines (TNF, IL-6, IL-1ß, CCL2, CXCL1, and CXCL2), an increased presence of CD68+ macrophages and an influx of neutrophils in the liver, and resulting apoptotic and necrotic hepatocyte cell death. Mechanistically, liver injury in cFLIP-/- mice was aggravated by reactive oxygen species, and sustained activation of the JNK signaling pathway. In parallel, cytoprotective NF-κB p65, A20, and the MAPK p38 were inhibited. Increased injury in cFLIP-/- mice was accompanied by activation of hepatic stellate cells and profibrogenic regulators. The antagonistic caspase 8-homolog cFLIP is a critical regulator of acute, cholestatic liver injury. NEW & NOTEWORTHY The current paper explores the role of a classical modulator of hepatocellular apoptosis in early, cholestatic liver injury. These include activation of NF-κB and MAPK signaling, production of inflammatory cytokines, and recruitment of neutrophils in response to cholestasis. Because these signaling pathways are currently exploited in clinical trials for the treatment of nonalcoholic steatohepatitis and cirrhosis, the current data will help in the development of novel pharmacological options in these indications.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Choledocholithiasis/metabolism , Common Bile Duct/surgery , Hepatic Stellate Cells/metabolism , Hepatitis/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Animals , Apoptosis , Bile Acids and Salts/metabolism , Bile Acids and Salts/toxicity , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cells, Cultured , Choledocholithiasis/etiology , Choledocholithiasis/genetics , Choledocholithiasis/pathology , Cytokines/metabolism , Genetic Predisposition to Disease , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology , Hepatitis/etiology , Hepatitis/genetics , Hepatitis/pathology , Hepatocytes/drug effects , Hepatocytes/pathology , Inflammation Mediators/metabolism , Ligation , Liver/drug effects , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice, Knockout , Necrosis , Neutrophil Infiltration , Oxidative Stress , Phenotype , Signal Transduction , Time Factors , Transcription Factor RelA/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Cell Signal ; 28(9): 1262-1269, 2016 09.
Article in English | MEDLINE | ID: mdl-27267061

ABSTRACT

Cellular-Flice-like inhibitory protein (c-FLIP) is an apoptosis modulator known to inhibit the extrinsic apoptotic pathway thus blocking Caspase-8 processing in the Death Inducing Signalling Complex (DISC). We previously demonstrated that c-FLIP localizes at the endoplasmic reticulum (ER) and that c-FLIP-deficient mouse embryonic fibroblasts (MEFs) display an enlarged ER morphology. In the present study, we have addressed the consequences of c-FLIP ablation in the ER stress response by investigating the effects of pharmacologically-induced ER stress in Wild Type (WT) and c-FLIP-/- MEFs. Surprisingly, c-FLIP-/- MEFs were found to be strikingly more resistant than WT MEFs to ER stress-mediated apoptosis. Analysis of Unfolded Protein Response (UPR) pathways revealed that Pancreatic ER Kinase (PERK) and Inositol-Requiring Enzyme 1 (IRE1) branch signalling is compromised in c-FLIP-/- cells when compared with WT cells. We found that c-FLIP modulates the PERK pathway by interfering with the activity of the serine threonine kinase AKT. Indeed, c-FLIP-/- MEFs display higher levels of active AKT than WT MEFs upon ER stress, while treatment with a specific AKT inhibitor of c-FLIP-/- MEFs subjected to ER stress restores the PERK but not the IRE1 pathway. Importantly, the AKT inhibitor or dominant negative AKT transfection sensitizes c-FLIP-/- cells to ER stress-induced cell death while the expression of a constitutively active AKT reduces WT cells sensitivity to ER stress-induced death. Thus, our results demonstrate that c-FLIP modulation of AKT activity is crucial in controlling PERK signalling and sensitivity to ER stress, and highlight c-FLIP as a novel molecular player in PERK and IRE1-mediated ER stress response.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Endoplasmic Reticulum Stress , Animals , Apoptosis , Autophagy , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Embryo, Mammalian/cytology , Enzyme Activation , Fibroblasts/metabolism , Membrane Proteins/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , eIF-2 Kinase/metabolism
4.
Chem Biol Interact ; 228: 69-78, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25619640

ABSTRACT

6-Shogaol, a potent bioactive compound in ginger (Zingiber officinale Roscoe), has been reported for anti-inflammatory and anti-cancer activity. In this study, we investigated the effect of 6-shogaol to enhance tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis. The combined treatment with 6-shogaol and TRAIL markedly induces apoptosis in various cancer cells (renal carcinoma Caki cells, breast carcinoma MDA-MB-231 cells and glioma U118MG cells), but not in normal mesangial cells and normal mouse kidney cells. 6-Shogaol reduced the mitochondrial membrane potential (MMP) and released cytochrome c from mitochondria to cytosol via Bax activation. Furthermore, we found that 6-shogaol induced down-regulation of c-FLIP(L) expression at the post-translational levels and the overexpression of c-FLIP(L) markedly inhibited 6-shogaol plus TRAIL-induced apoptosis. Moreover, 6-shogaol increased reactive oxygen species (ROS) production in Caki cells. Pretreatment with ROS scavengers attenuated 6-shogaol plus TRAIL-induced apoptosis through inhibition of MMP reduction and down-regulation of c-FLIP(L) expression. In addition, 6-gingerol, another phenolic alkanone isolated from ginger, did not enhance TRAIL-induced apoptosis and down-regulate c-FLIP(L) expression. Taken together, our results demonstrated that 6-shogaol enhances TRAIL-mediated apoptosis in renal carcinoma Caki cells via ROS-mediated cytochrome c release and down-regulation of c-FLIP(L) expression.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Carcinoma, Renal Cell/drug therapy , Catechols/pharmacology , Cytochromes c/metabolism , Kidney Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Catechols/chemistry , Catechols/isolation & purification , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Structure-Activity Relationship
5.
Cell Rep ; 5(2): 397-408, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24209745

ABSTRACT

FADD, caspase-8, and cFLIP regulate the outcome of cell death signaling. Mice that constitutively lack these molecules die at an early embryonic age, whereas tissue-specific constitutive deletion of FADD or caspase-8 results in inflammatory skin disease caused by increased necroptosis. The function of cFLIP in the skin in vivo is unknown. In contrast to tissue-specific caspase-8 knockout, we show that mice constitutively lacking cFLIP in the epidermis die around embryonic days 10 and 11. When cFLIP expression was abrogated in adult skin of cFLIPfl/fl-K14CreERtam mice, severe inflammation of the skin with concomitant caspase activation and apoptotic, but not necroptotic, cell death developed. Apoptosis was dependent of autocrine tumor necrosis factor production triggered by loss of cFLIP. In addition, epidermal cFLIP protein was lost in patients with severe drug reactions associated with epidermal apoptosis. Our data demonstrate the importance of cFLIP for the integrity of the epidermis and for silencing of spontaneous skin inflammation.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Keratinocytes/metabolism , Skin/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Caspase 8/metabolism , Cells, Cultured , Homeostasis , Keratinocytes/cytology , Keratinocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/pathology
6.
Cell Rep ; 5(2): 340-8, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24095739

ABSTRACT

Caspase-8 or cellular FLICE-like inhibitor protein (cFLIP) deficiency leads to embryonic lethality in mice due to defects in endothelial tissues. Caspase-8(-/-) and receptor-interacting protein kinase-3 (RIPK3)(-/-), but not cFLIP(-/-) and RIPK3(-/-), double-knockout animals develop normally, indicating that caspase-8 antagonizes the lethal effects of RIPK3 during development. Here, we show that the acute deletion of caspase-8 in the gut of adult mice induces enterocyte death, disruption of tissue homeostasis, and inflammation, resulting in sepsis and mortality. Likewise, acute deletion of caspase-8 in a focal region of the skin induces local keratinocyte death, tissue disruption, and inflammation. Strikingly, RIPK3 ablation rescues both phenotypes. However, acute loss of cFLIP in the skin produces a similar phenotype that is not rescued by RIPK3 ablation. TNF neutralization protects from either acute loss of caspase-8 or cFLIP. These results demonstrate that caspase-8-mediated suppression of RIPK3-induced death is required not only during development but also for adult homeostasis. Furthermore, RIPK3-dependent inflammation is dispensable for the skin phenotype.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Animals , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Enterocytes/cytology , Homeostasis/drug effects , Mice , Mice, Knockout , Neutrophils/metabolism , Neutrophils/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Skin/drug effects , Skin/metabolism , Tamoxifen/pharmacology , Tumor Necrosis Factor-alpha/metabolism
7.
Gastroenterology ; 145(6): 1369-79, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24036366

ABSTRACT

BACKGROUND & AIMS: The intestinal epithelium generates a barrier that protects mammals from potentially harmful intestinal contents, such as pathogenic bacteria. Dysregulation of epithelial cell death has been implicated in barrier dysfunction and in the pathogenesis of intestinal inflammation. We investigated mechanisms of cell-death regulation in the intestinal epithelium of mice. METHODS: Conditional knockout mice (either inducible or permanent) with deletion of cellular FLICE-inhibitory protein (cFlip) or caspase-8 in the intestinal epithelium were analyzed by histology and high-resolution endoscopy. We assessed the effects of cFlip or caspase-8 deficiency on intestinal homeostasis. RESULTS: Expression of cFlip in the intestinal epithelium was required for constitutive activation of caspase-8 under steady-state conditions. Intestinal expression of cFlip was required for development; disruption of the gene encoding cFlip from the intestinal epithelium (cFlip(fl/fl) VillinCre(+) mice) resulted in embryonic lethality. When cFlip was deleted from the intestinal epithelium of adult mice (cFlip(iΔIEC) mice), the animals died within a few days from severe tissue destruction, epithelial cell death, and intestinal inflammation. Death of cFlip-depleted intestinal epithelial cells was regulated extrinsically and required the presence of death receptor ligands, such as tumor necrosis factor-α and CD95 ligand, but was independent of receptor-interacting protein 3. cFlip deficiency was associated with strong up-regulation of caspase-8 and caspase-3 activity and excessive apoptosis in intestinal crypts. CONCLUSIONS: cFlip is required for intestinal tissue homeostasis in mice. It controls the level of activation of caspase-8 to promote survival of intestinal epithelial cells.


Subject(s)
Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Caspase 8/physiology , Homeostasis/physiology , Immunity/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 3/physiology , Cell Survival/physiology , Female , Male , Mice , Mice, Knockout , Microfilament Proteins/physiology , Models, Animal , Signal Transduction/physiology , Up-Regulation/physiology
8.
Am J Physiol Gastrointest Liver Physiol ; 303(4): G498-506, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22700824

ABSTRACT

Chronic liver disease promotes hepatocellular injury involving apoptosis and triggers compensatory regeneration that leads to the activation of quiescent stellate cells in the liver. The deposition of extracellular matrix from activated myofibroblasts promotes hepatic fibrosis and the progression to cirrhosis with deleterious effects on liver physiology. The role of apoptosis signaling pathways in the development of fibrosis remains undefined. The aim of the current study was to determine the involvement of the caspase-8 homologue cellular FLICE-inhibitory protein (cFLIP) during the initiation and progression of fibrosis. Liver injury and fibrosis from carbon tetrachloride (CCl(4)) and thioacetamide (TAA) were examined in mice exhibiting a hepatocyte-specific deletion of cFLIP (flip(-/-)). Acute liver injury from CCl(4) and TAA were enhanced in flip(-/-) mice. This was accompanied by increased activation of caspase-3 and -9, pronounced phosphorylation of JNK, and decreased phosphorylation of Erk. Deletion of the cJun NH(2)-terminal kinase 2 (JNK2) in flip(-/-) mice protected from injury. Hepatic fibrosis was increased at baseline in 12-wk-old flip(-/-) mice, and progression of fibrosis from TAA was accelerated compared with the wild type. In conclusion, deletion of cFLIP in hepatocytes leads to increased fibrosis and accelerated fibrosis progression. This is accompanied by increased injury involving the activation of caspases and JNK2. Thus predisposition to liver injury involving increased hepatocellular apoptosis is a critical mediator of accelerated fibrogenesis, and prevention of liver injury will be a most important measure for patients with chronic liver disease.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/enzymology , Liver Cirrhosis/etiology , Liver/enzymology , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Carbon Tetrachloride , Caspase 3/metabolism , Caspase 9/metabolism , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Disease Progression , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Genotype , Hepatocytes/pathology , Liver/pathology , Liver Cirrhosis/enzymology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Phenotype , Phosphorylation , Signal Transduction , Thioacetamide , Time Factors
9.
Cell Rep ; 1(5): 401-7, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22675671

ABSTRACT

Caspase-8, the initiator caspase of the death receptor pathway of apoptosis, its adapter molecule, FADD, required for caspase-8 activation, and cFLIPL, a caspase-8-like protein that lacks a catalytic site and blocks caspase-8-mediated apoptosis, are each essential for embryonic development. Animals deficient in any of these genes present with E10.5 embryonic lethality. Recent studies have shown that development in caspase-8-deficient mice is rescued by ablation of RIPK3, a kinase that promotes a form of programmed, necrotic cell death. Here, we show that FADD, RIPK3 double-knockout mice develop normally but that the lethal effects of cFLIP deletion are not rescued by RIPK3 deficiency. Remarkably, in mice lacking FADD, cFLIP, and RIPK3, embryonic development is normal. This can be explained by the convergence of two cell processes: the enzymatic activity of the FADD-caspase-8-cFLIPL complex blocks RIPK3-dependent signaling (including necrosis), whereas cFLIPL blocks RIPK3-independent apoptosis promoted by the FADD-caspase-8 complex.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Caspase 8/physiology , Embryonic Development/physiology , Fas-Associated Death Domain Protein/physiology , Signal Transduction/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Embryonic Development/genetics , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Gene Deletion , Mice , Mice, Knockout , Necrosis/genetics , Necrosis/physiopathology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Signal Transduction/genetics
10.
J Hepatol ; 55(6): 1272-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21703207

ABSTRACT

BACKGROUND & AIMS: Apoptosis is crucially involved in acute and chronic liver injury, including viral, cholestatic, toxic, and metabolic liver disease. Additionally, dysregulation of apoptosis signaling pathways has been implicated in hepatocarcinogenesis. The most prominent members of the apoptosis-mediating tumor necrosis factor receptor superfamily are the TNF-R1 (CD120a) and the CD95 (Apo-1/Fas) receptor. Although extensively studied, the intracellular signaling events in hepatocytes are only incompletely understood. METHODS: To examine the role of the caspase-8 homolog cellular FLICE-inhibitory protein (c-FLIP) in liver injury, we generated mice with hepatocyte specific deletion of c-FLIP. Three models of acute liver injury were employed: the agonistic anti-CD95 antibody Jo2, d-galactosamine and LPS (GalN/LPS), and concanavalin A. RESULTS: Conditional ablation of c-FLIP in hepatocytes augmented liver injury and cell death in all three models of liver injury. CD95- and GalN/LPS-induced liver injury was ameliorated by a pancaspase inhibitor, while ConA-induced injury was unaffected by caspase inhibition. Augmented activation of the MAPK JNK was observed in parallel to liver injury in c-FLIP knockout mice in all injury models; however, inhibition of JNK only affected TNF- and ConA-mediated injury. CONCLUSIONS: In summary, c-FLIP is a central regulator of cell death in hepatocytes, involving increased activation of caspases and the MAPK JNK.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Receptors, Death Domain/metabolism , Animals , Anthracenes/pharmacology , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspases/metabolism , Concanavalin A/toxicity , Female , Galactosamine/toxicity , Hepatocytes/drug effects , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , fas Receptor/agonists
11.
Exp Cell Res ; 317(13): 1841-50, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21621534

ABSTRACT

TNFα stimulation triggers both cell death and survival programs. Since dysregulated apoptosis or cell growth can cause inflammatory diseases, cancer, or autoimmune disorders, it is important to understand the molecular mechanism of controlling cell death and survival by TNFR downstream signaling molecules. In this study, we used normal diploid cells, mouse embryonic fibroblasts (MEFs), to mimic the general TNFα-resistant phenomenon seen under physiological conditions.We elucidated the TNFα-induced death signaling complexes in TNF α-resistant WT MEFs and TNFα-sensitive MEFs that were cFLIP-, RelA-, TRAF2- or RIP1-deficient. Consistent with TNFα-mediated killing, we detected TNFα-induced high molecular weight complexes containing caspase-8 and FADD by gel filtration in the deficient MEFs, especially in those devoid of cFLIP. In addition to the presence of caspase-8-FADD in the TNFα-induced-death complex in the deficient MEFs, we also detected an intermediate protein complex containing RIP1, TRAF2 and caspase-8.Moreover, we demonstrated a correlation between TNFα-sensitivity and death-inducing complex ability in two transformed cell lines, E1A- and Ras- transformed MEFs and PDGF-B-transformed NIH-3T3 cells with PDGF-B signaling inhibited by the tyrosine kinase inhibitor STI571. Taken together, our results suggest the involvement of cFLIP-, RelA-, RIP1-, or TRAF2-related mechanisms for preventing FADD-caspase-8 interaction in wild-type MEFs.


Subject(s)
Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , Caspase 8/metabolism , Cell Death , Cells, Cultured , Embryo, Mammalian/cytology , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , GTPase-Activating Proteins/deficiency , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , NIH 3T3 Cells , TNF Receptor-Associated Factor 2/deficiency , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
12.
Blood ; 117(2): 618-29, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20980680

ABSTRACT

The timely clearance of apoptotic neutrophils from inflammation sites is an important function of macrophages; however, the role of macrophages in maintaining neutrophil homeostasis under steady-state conditions is less well understood. By conditionally deleting the antiapoptotic gene cellular FLICE-like inhibitory protein (C-FLIP) in myeloid cells, we have generated a novel mouse model deficient in marginal zone and bone marrow stromal macrophages. These mice develop severe neutrophilia, splenomegaly, extramedullary hematopoiesis, decreased body weight, and increased production of granulocyte colony-stimulating factor (G-CSF) and IL-1ß, but not IL-17. c-FLIP(f/f) LysM-Cre mice exhibit delayed clearance of circulating neutrophils, suggesting that failure of macrophages to efficiently clear apoptotic neutrophils causes production of cytokines that drive excess granulopoiesis. Further, blocking G-CSF but not IL-1R signaling in vivo rescues this neutrophilia, suggesting that a G-CSF-dependent, IL-1ß-independent pathway plays a role in promoting neutrophil production in mice with defective clearance of apoptotic cells.


Subject(s)
Homeostasis/immunology , Macrophages/immunology , Neutrophils/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Survival , Gene Knock-In Techniques , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoiesis/genetics , Hematopoiesis/immunology , Homeostasis/genetics , Inflammation/genetics , Inflammation/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neutrophils/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism
13.
J Immunol ; 184(9): 4871-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20335528

ABSTRACT

High levels of the Fas-signaling antagonist cellular FLIP (cFLIP) in germinal center (GC) B cells suggests an important role for this factor during this stage of the T cell-dependent B cell immune response. To test this idea, we used mice with B cell-specific deletion of a floxed cFLIP allele. Although deletion of cFLIP did not alter their primary development, participation of cFLIP-deficient B cells in the immune response was severely perturbed. Using previously characterized IgH locus-targeted BCR transgenic mice, we showed that adoptively transferred cFLIP-deficient follicular B cells do not effectively participate in the GC response in wild-type hosts. However, this failure was accompanied by severe defects in the initial activation and proliferation of these B cells in vivo. In addition, immunization of mice with B cell-specific cFLIP deletion resulted in selective recruitment into GCs and Ab-forming cell responses of B cells that had not deleted the floxed cFLIP allele. Together, these findings demonstrate that expression of cFLIP is a prerequisite for participation of B cells in all stages of Ag-driven immune responses.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Gene Expression Regulation/immunology , Germinal Center/immunology , Adoptive Transfer , Alleles , Animals , Antigens, CD19/genetics , Antigens, CD19/physiology , B-Lymphocyte Subsets/transplantation , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Gene Deletion , Gene Knock-In Techniques , Germinal Center/cytology , Germinal Center/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/physiology
14.
Gynecol Oncol ; 117(3): 451-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20227749

ABSTRACT

OBJECTIVE: In the current study, we aimed to investigate the role of the long isoform of cellular Fas-associated death domain-like interleukin-1beta-converting enzyme (FLICE)-like inhibitory protein (c-FLIP(L)) in ovarian cancer (OC) development by using RNA interference (RNAi) in vitro and in vivo. METHODS: TRAIL-resistant human OC cell lines were genetically manipulated by RNAi-mediated suppression of c-FLIP(L). Subsequently, the genetic alteration that was introduced into the various OC cell lines was characterized in vitro and in vivo. RESULTS: We previously showed that about 40% of OC patients express high levels of c-FLIP(L), and that natural killer (NK) cells mediated immunosurveillance in OC. In the present study, we observed that the knockdown of c-FLIP(L) in human OC cell lines not only enhanced their sensitivity to TRAIL-mediated apoptosis, but also inhibited their migratory phenotype in a TRAIL-dependent manner in vitro. Shutdown of c-FLIP(L) in OC cells significantly decreased tumor development by induction of apoptosis and reduction of proliferation in vivo. Importantly, the knockdown of c-FLIP(L) particularly inhibited the invasion of OC cells into the peritoneal cavity, which might be due to high expression of TRAIL by NK cells and NK-cell mediated immunosurveillance. CONCLUSION: These data demonstrate that c-FLIP(L) exhibits multiple functions in OC cells: first by concomitantly evading the natural immunity mediated by TRAIL-induced cell death, and second by augmenting cell motility and invasion in vivo. Our findings indicate that c-FLIP(L) regulates sensitivity of OC to TRAIL-mediated apoptosis and offers possible therapeutical implications for OC in the future.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Animals , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/immunology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Down-Regulation , Drug Resistance, Neoplasm , Female , Gene Knockdown Techniques , Humans , Immunologic Surveillance , Mice , Mice, Nude , Neoplasm Invasiveness , Ovarian Neoplasms/drug therapy , RNA, Small Interfering/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Xenograft Model Antitumor Assays
15.
J Immunol ; 182(1): 207-15, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109151

ABSTRACT

Fas/Apo-1 signals through the FADD (Fas-associated death domain) adaptor protein, which recruits and activates the apical caspase 8 and leads to apoptosis. Cellular FLIP (cFLIP) is a homolog of caspase 8 and is also capable of binding to FADD. Previous studies suggest that cFLIP could either enhance or inhibit apoptosis and lead to NF-kappaB and Erk1/2 activation. Like FADD or caspase 8 deficiency, a lack of cFLIP disrupts embryogenesis and T cell proliferation. It has been demonstrated that B cells lacking either FADD or caspase 8 were defective in both Fas-induced apoptosis and TLR-induced proliferation, which indicates that these death-inducing proteins have an additional role in regulating innate immunity. To analyze the function of cFLIP in B cells, conditional deletion of cFLIP was induced by using CD19(Cre). The resulting B cell-specific cFLIP-deficient mice were found to have reduced numbers of peripheral B cells that were hypersensitive to Fas-induced apoptosis and impaired in proliferation induced by TLRs and the BCR. Furthermore, there was aberrant expression of costimulatory proteins and activation markers in cFLIP-deficient B cells. Whereas LPS-induced activation of NF-kappaB and Erk1/2 appears to be unaffected, p38 and Jnk were spontaneously activated and hyperinduced in cFLIP-deficient B cells. Therefore, these data revealed novel functions of cFLIP in B cells.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/enzymology , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Cell Proliferation , Mitogen-Activated Protein Kinase 9/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/immunology , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/immunology , Mice , Mice, Knockout , Mice, Transgenic
16.
Oncogene ; 27(1): 76-84, 2008 Jan 03.
Article in English | MEDLINE | ID: mdl-17599041

ABSTRACT

Nuclear factor-kappa B (NF-kappaB) inhibits cell death through suppression of the caspase cascade, the c-Jun N-terminal kinase (JNK) pathway, and reactive oxygen species (ROS) accumulation. To suppress this antiapoptotic function of NF-kappaB might be a promising strategy to increase susceptibility of tumor cells to stress-induced cell death. We have recently shown that tumor necrosis factor (TNF)alpha induces caspase-dependent and -independent JNK activation and ROS accumulation in cellular FLICE-inhibitory protein (c-Flip)(-/-) murine embryonic fibroblasts (MEFs). To apply this observation to tumor therapy, we knocked down c-FLIP by RNA interference in various tumor cells. Consistent with the results using c-Flip(-/-) MEFs, we found that TNFalpha stimulation induced caspase-dependent prolonged JNK activation and ROS accumulation, followed by apoptotic and necrotic cell death in various tumor cells. Furthermore, TNFalpha and Fas induced the cleavage of mitogen-activated protein kinase/ERK kinase kinase (MEKK)1, resulting in generation of a constitutive active form of MEKK1 leading to JNK activation in c-FLIP knockdown cells. Given that ROS accumulation and necrotic cell death enhance inflammation followed by compensatory proliferation of tumor cells, selective suppression of caspase-dependent ROS accumulation will be an alternative strategy to protect cells from ROS-dependent DNA damage and compensatory tumor progression.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/antagonists & inhibitors , Caspases/physiology , Down-Regulation/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Uterine Cervical Neoplasms/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Enzyme Activation/physiology , Female , HCT116 Cells , HeLa Cells , Humans , RNA, Small Interfering/genetics , Tumor Necrosis Factor-alpha/physiology , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...